Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 155
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Gut ; 70(3): 555-566, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32641470

RESUMO

OBJECTIVE: Patients with Lynch syndrome (LS) are at markedly increased risk for colorectal cancer. It is being increasingly recognised that the immune system plays an essential role in LS tumour development, thus making an ideal target for cancer prevention. Our objective was to evaluate the safety, assess the activity and discover novel molecular pathways involved in the activity of naproxen as primary and secondary chemoprevention in patients with LS. DESIGN: We conducted a Phase Ib, placebo-controlled, randomised clinical trial of two dose levels of naproxen sodium (440 and 220 mg) administered daily for 6 months to 80 participants with LS, and a co-clinical trial using a genetically engineered mouse model of LS and patient-derived organoids (PDOs). RESULTS: Overall, the total number of adverse events was not different across treatment arms with excellent tolerance of the intervention. The level of prostaglandin E2 in the colorectal mucosa was significantly decreased after treatment with naproxen when compared with placebo. Naproxen activated different resident immune cell types without any increase in lymphoid cellularity, and changed the expression patterns of the intestinal crypt towards epithelial differentiation and stem cell regulation. Naproxen demonstrated robust chemopreventive activity in a mouse co-clinical trial and gene expression profiles induced by naproxen in humans showed perfect discrimination of mice specimens with LS and PDOs treated with naproxen and control. CONCLUSIONS: Naproxen is a promising strategy for immune interception in LS. We have discovered naproxen-induced gene expression profiles for their potential use as predictive biomarkers of drug activity. TRIAL REGISTRATION NUMBER: gov Identifier: NCT02052908.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Quimioprevenção , Neoplasias Colorretais Hereditárias sem Polipose/tratamento farmacológico , Neoplasias Colorretais Hereditárias sem Polipose/imunologia , Naproxeno/farmacologia , Adulto , Idoso , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Dinoprostona/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Mucosa Intestinal/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Naproxeno/administração & dosagem
3.
Carcinogenesis ; 36(2): 291-8, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25503930

RESUMO

Non-steroidal anti-inflammatory drugs prevent colorectal cancer by inhibiting cyclooxygenase (COX) enzymes that synthesize tumor-promoting prostaglandins. 15-hydroxyprostaglandin dehydrogenase (15-PGDH) is a tumor suppressor that degrades tumor-promoting prostaglandins. Murine knockout of 15-PGDH increases susceptibility to azoxymethane-induced colon tumors. It also renders these mice resistant to celecoxib, a selective inhibitor of inducible COX-2 during colon neoplasia. Similarly, humans with low colonic 15-PGDH are also resistant to colon adenoma prevention with celecoxib. Here, we used aspirin and sulindac, which inhibit both COX-1 and COX-2, in order to determine if these broader COX inhibitors can prevent colon tumors in 15-PGDH knockout (KO) mice. Unlike celecoxib, sulindac proved highly effective in colon tumor prevention of 15-PGDH KO mice. Significantly, however, aspirin demonstrated no effect on colon tumor incidence in either 15-PGDH wild-type or KO mice, despite a comparable reduction in colonic mucosal Prostaglandin E2 (PGE2) levels by both sulindac and aspirin. Notably, colon tumor prevention activity by sulindac was accompanied by a marked induction of lymphoid aggregates and proximal colonic inflammatory mass lesions, a side effect seen to a lesser degree with celecoxib, but not with aspirin. These findings suggest that sulindac may be the most effective agent for colon cancer prevention in humans with low 15-PGDH, but its use may also be associated with inflammatory lesions in the colon.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/prevenção & controle , Hidroxiprostaglandina Desidrogenases/genética , Sulindaco/farmacologia , Animais , Antineoplásicos/farmacologia , Aspirina/farmacologia , Azoximetano , Carcinógenos , Celecoxib , Quimioprevenção , Neoplasias do Colo/induzido quimicamente , Ciclo-Oxigenase 1/efeitos dos fármacos , Ciclo-Oxigenase 2/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase 2/farmacologia , Dinoprostona/metabolismo , Inflamação/imunologia , Mucosa Intestinal/patologia , Proteínas de Membrana/efeitos dos fármacos , Camundongos , Camundongos Knockout , Pirazóis/farmacologia , Sulfonamidas/farmacologia
4.
J Biol Chem ; 289(10): 6513-6525, 2014 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-24451373

RESUMO

The p53 tumor suppressor gene encodes a homotetrameric transcription factor which is activated in response to a variety of cellular stressors, including DNA damage and oncogene activation. p53 mutations occur in >50% of human cancers. Although p53 has been shown to regulate Wnt signaling, the underlying mechanisms are not well understood. Here we show that silencing p53 in colon cancer cells led to increased expression of Aha1, a co-chaperone of Hsp90. Heat shock factor-1 was important for mediating the changes in Aha1 levels. Increased Aha1 levels were associated with enhanced interactions with Hsp90, resulting in increased Hsp90 ATPase activity. Moreover, increased Hsp90 ATPase activity resulted in increased phosphorylation of Akt and glycogen synthase kinase-3ß (GSK3ß), leading to enhanced expression of Wnt target genes. Significantly, levels of Aha1, Hsp90 ATPase activity, Akt, and GSK3ß phosphorylation and expression of Wnt target genes were increased in the colons of p53-null as compared with p53 wild type mice. Using p53 heterozygous mutant epithelial cells from Li-Fraumeni syndrome patients, we show that a monoallelic mutation of p53 was sufficient to activate the Aha1/Hsp90 ATPase axis leading to stimulation of Wnt signaling and increased expression of Wnt target genes. Pharmacologic intervention with CP-31398, a p53 rescue agent, inhibited recruitment of Aha1 to Hsp90 and suppressed Wnt-mediated gene expression in colon cancer cells. Taken together, this study provides new insights into the mechanism by which p53 regulates Wnt signaling and raises the intriguing possibility that p53 status may affect the efficacy of anticancer therapies targeting Hsp90 ATPase.


Assuntos
Adenosina Trifosfatases/metabolismo , Neoplasias do Colo/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas de Choque Térmico HSP90/metabolismo , Chaperonas Moleculares/genética , Proteína Supressora de Tumor p53/metabolismo , Via de Sinalização Wnt/genética , Animais , Neoplasias do Colo/genética , Modelos Animais de Doenças , Humanos , Síndrome de Li-Fraumeni , Camundongos , Camundongos Transgênicos , Pirimidinas/farmacologia , Proteína Supressora de Tumor p53/genética
5.
J Biol Chem ; 288(33): 23858-67, 2013 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-23839987

RESUMO

Methylation of cytosines in CpG dinucleotides is the predominant epigenetic mark on vertebrate DNA. DNA methylation is associated with transcriptional repression. The pattern of DNA methylation changes during development and with disease. Human DNA methyltransferase 1 (Dnmt1), a 1616-amino acid multidomain enzyme, is essential for maintenance of DNA methylation in proliferating cells and is considered an important cancer drug target. Using a fluorogenic, endonuclease-coupled DNA methylation assay with an activated form of Dnmt1 engineered to lack the replication foci targeting sequence domain, we discovered that laccaic acid A (LCA), a highly substituted anthraquinone natural product, is a direct inhibitor with a 310 nm Ki. LCA is competitive with the DNA substrate in in vitro methylation assays and alters the expression of methylated genes in MCF-7 breast cancer cells synergistically with 5-aza-2'-deoxycytidine. LCA represents a novel class of Dnmt-targeted molecular probes, with biochemical properties that allow it to distinguish between non DNA-bound and DNA-bound Dnmt1.


Assuntos
Compostos Azo/farmacologia , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , DNA/metabolismo , Inibidores Enzimáticos/farmacologia , Aminoquinolinas/química , Aminoquinolinas/farmacologia , Antraquinonas/farmacologia , Azacitidina/análogos & derivados , Azacitidina/química , Azacitidina/farmacologia , Compostos Azo/química , Sequência de Bases , Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/genética , Replicação do DNA/efeitos dos fármacos , Decitabina , Inibidores Enzimáticos/química , Feminino , Fluorometria , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células MCF-7 , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Pirimidinas/química , Pirimidinas/farmacologia , Reprodutibilidade dos Testes , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Temperatura de Transição
6.
Avian Dis ; 58(1): 95-101, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24758120

RESUMO

There is a paucity of preclinical models that simulate the development of ovarian tumors in humans. At present, the egg-laying hen appears to be the most promising model to study the spontaneous occurrence of ovarian tumors in the clinical setting. Although gross classification and histologic grade of tumors have been used prognostically in women with ovarian tumors, there is currently no single system that is universally used to classify reproductive tumors in the hen. Four hundred and one 192-wk-old egg-laying hens were necropsied to determine the incidence of reproductive tumors using both gross pathology and histologic classification. Gross pathologic classifications were designated as follows: birds presenting with ovarian tumors only (class 1), those presenting with oviductal and ovarian tumors (class 2), those with ovarian and oviductal tumors that metastasized to the gastrointestinal tract (class 3), those with ovarian and oviductal tumors that metastasized to the gastrointestinal tract and other distant organs (class 4), those with oviductal tumors only (class 5), those with oviductal tumors that metastasized to other organs with no ovarian involvement (class 6), and those with ovarian tumors that metastasized to other organs with no oviductal involvement (class 7), including birds with gastrointestinal tumors and no reproductive involvement (GI only) and those with no tumors (normal). Histopathologic classifications range from grades 1 to 3 and are based on mitotic developments and cellular differentiation. An updated gross pathology and histologic classification systems for the hen reproductive malignancies provides a method to report the range of reproductive tumors revealed in a flock of aged laying hens.


Assuntos
Galinhas , Células Epiteliais/patologia , Neoplasias Gastrointestinais/veterinária , Doenças dos Genitais Femininos/veterinária , Neoplasias Ovarianas/veterinária , Oviductos/patologia , Doenças das Aves Domésticas/patologia , Animais , Feminino , Neoplasias Gastrointestinais/classificação , Neoplasias Gastrointestinais/epidemiologia , Neoplasias Gastrointestinais/patologia , Doenças dos Genitais Femininos/classificação , Doenças dos Genitais Femininos/epidemiologia , Doenças dos Genitais Femininos/patologia , Incidência , Neoplasias Ovarianas/classificação , Neoplasias Ovarianas/epidemiologia , Neoplasias Ovarianas/patologia , Doenças das Aves Domésticas/classificação , Doenças das Aves Domésticas/epidemiologia
7.
Proc Natl Acad Sci U S A ; 108(6): 2444-9, 2011 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-21248225

RESUMO

Cancer biomarkers are currently the subject of intense research because of their potential utility for diagnosis, prognosis, and targeted therapy. In theory, the gene products resulting from somatic mutations are the ultimate protein biomarkers, being not simply associated with tumors but actually responsible for tumorigenesis. We show here that the altered protein products resulting from somatic mutations can be identified directly and quantified by mass spectrometry. The peptides expressed from normal and mutant alleles were detected by selected reaction monitoring (SRM) of their product ions using a triple-quadrupole mass spectrometer. As a prototypical example of this approach, we demonstrated that it is possible to quantify the number and fraction of mutant Ras protein present in cancer cell lines. There were an average of 1.3 million molecules of Ras protein per cell, and the ratio of mutant to normal Ras proteins ranged from 0.49 to 5.6. Similarly, we found that mutant Ras proteins could be detected and quantified in clinical specimens such as colorectal and pancreatic tumor tissues as well as in premalignant pancreatic cyst fluids. In addition to answering basic questions about the relative levels of genetically abnormal proteins in tumors, this approach could prove useful for diagnostic applications.


Assuntos
Biomarcadores Tumorais/metabolismo , Proteínas Mutantes/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Proteínas ras/metabolismo , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Feminino , Humanos , Masculino , Proteínas Mutantes/genética , Proteínas de Neoplasias/genética , Neoplasias/genética , Proteínas ras/genética
8.
Proc Natl Acad Sci U S A ; 108(52): 21188-93, 2011 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-22158988

RESUMO

More than 2% of adults harbor a pancreatic cyst, a subset of which progresses to invasive lesions with lethal consequences. To assess the genomic landscapes of neoplastic cysts of the pancreas, we determined the exomic sequences of DNA from the neoplastic epithelium of eight surgically resected cysts of each of the major neoplastic cyst types: serous cystadenomas (SCAs), intraductal papillary mucinous neoplasms (IPMNs), mucinous cystic neoplasms (MCNs), and solid pseudopapillary neoplasms (SPNs). SPNs are low-grade malignancies, and IPMNs and MCNs, but not SCAs, have the capacity to progress to cancer. We found that SCAs, IPMNs, MCNs, and SPNs contained 10 ± 4.6, 27 ± 12, 16 ± 7.6, and 2.9 ± 2.1 somatic mutations per tumor, respectively. Among the mutations identified, E3 ubiquitin ligase components were of particular note. Four of the eight SCAs contained mutations of the von Hippel-Lindau gene (VHL), a key component of the VHL ubiquitin ligase complex that has previously been associated with renal cell carcinomas, SCAs, and other neoplasms. Six of the eight IPMNs and three of the eight MCNs harbored mutations of RNF43, a gene coding for a protein with intrinsic E3 ubiquitin ligase activity that has not previously been found to be genetically altered in any human cancer. The preponderance of inactivating mutations in RNF43 unequivocally establish it as a suppressor of both IPMNs and MCNs. SPNs contained remarkably few genetic alterations but always contained mutations of CTNNB1, previously demonstrated to inhibit degradation of the encoded protein (ß-catenin) by E3 ubiquitin ligases. These results highlight the essential role of ubiquitin ligases in these neoplasms and have important implications for the diagnosis and treatment of patients with cystic tumors.


Assuntos
Cistadenoma Mucinoso/genética , Cistadenoma Seroso/genética , Exoma/genética , Mutação/genética , Cisto Pancreático/genética , Papiloma Intraductal/genética , Adulto , Sequência de Bases , Análise Mutacional de DNA , Proteínas de Ligação a DNA/genética , Humanos , Dados de Sequência Molecular , Proteínas Oncogênicas/genética , Cisto Pancreático/patologia , Ubiquitina-Proteína Ligases/genética , Proteína Supressora de Tumor Von Hippel-Lindau/genética
9.
J Proteome Res ; 12(10): 4351-65, 2013 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-24004147

RESUMO

Plasma proteomic experiments performed rapidly and economically using several of the latest high-resolution mass spectrometers were compared. Four quantitative hyperfractionated plasma proteomics experiments were analyzed in replicates by two AB SCIEX TripleTOF 5600 and three Thermo Scientific Orbitrap (Elite/LTQ-Orbitrap Velos/Q Exactive) instruments. Each experiment compared two iTRAQ isobaric-labeled immunodepleted plasma proteomes, provided as 30 labeled peptide fractions, and 480 LC-MS/MS runs delivered >250 GB of data in 2 months. Several analysis algorithms were compared. At 1% false discovery rate, the relative comparative findings concluded that the Thermo Scientific Q Exactive Mass Spectrometer resulted in the highest number of identified proteins and unique sequences with iTRAQ quantitation. The confidence of iTRAQ fold-change for each protein is dependent on the overall ion statistics (Mascot Protein Score) attainable by each instrument. The benchmarking also suggested how to further improve the mass spectrometry parameters and HPLC conditions. Our findings highlight the special challenges presented by the low abundance peptide ions of iTRAQ plasma proteome because the dynamic range of plasma protein abundance is uniquely high compared with cell lysates, necessitating high instrument sensitivity.


Assuntos
Proteínas Sanguíneas/química , Espectrometria de Massas em Tandem/métodos , Proteínas Sanguíneas/isolamento & purificação , Proteínas Sanguíneas/metabolismo , Humanos , Imunoprecipitação , Mapeamento de Peptídeos , Proteômica , Padrões de Referência , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Espectrometria de Massas em Tandem/instrumentação , Espectrometria de Massas em Tandem/normas
10.
Biochem Biophys Res Commun ; 435(4): 557-61, 2013 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-23665330

RESUMO

Rhabdomyosarcomas (RMS) represent the most common childhood soft-tissue sarcoma. Over the past few decades outcomes for low and intermediate risk RMS patients have slowly improved while patients with metastatic or relapsed RMS still face a grim prognosis. New chemotherapeutic agents or combinations of chemotherapies have largely failed to improve the outcome. Based on the identification of novel molecular targets, potential therapeutic approaches in RMS may offer a decreased reliance on conventional chemotherapy. Thus, identification of effective therapeutic agents that specifically target relevant pathways may be particularly beneficial for patients with metastatic and refractory RMS. The PI3K/AKT/mTOR pathway has been found to be a potentially attractive target in RMS therapy. In this study, we provide evidence that rapamycin (sirolimus) abrogates growth of RMS development in a RMS xenograft mouse model. As compared to a vehicle-treated control group, more than 95% inhibition in tumor growth was observed in mice receiving parenteral administration of rapamycin. The residual tumors in rapamycin-treated group showed significant reduction in the expression of biomarkers indicative of proliferation and tumor invasiveness. These tumors also showed enhanced apoptosis. Interestingly, the mechanism by which rapamycin diminished RMS tumor growth involved simultaneous inhibition of mTOR and hedgehog (Hh) pathways. Diminution in these pathways in this model of RMS also inhibited epithelial mesenchymal transition (EMT) which then dampened the invasiveness of these tumors. Our data provide bases for using rapamycin either alone or in combination with traditional chemotherapeutic drugs to block the pathogenesis of high risk RMS.


Assuntos
Rabdomiossarcoma/patologia , Rabdomiossarcoma/terapia , Sirolimo/uso terapêutico , Serina-Treonina Quinases TOR/metabolismo , Animais , Antibióticos Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Camundongos , Camundongos Nus , Transdução de Sinais/efeitos dos fármacos , Resultado do Tratamento
11.
Toxicol Appl Pharmacol ; 268(3): 249-55, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23274568

RESUMO

Nitric oxide (NO)-releasing non-steroidal anti-inflammatory drugs (NO-NSAIDs) which have been synthesized to reduce gastro-intestinal and cardiovascular toxicities of NSAIDs, possess anti-proliferative, pro-apoptotic and anti-cancer activities. Here, we show that NO-sulindac inhibited UVB-induced skin tumorigenesis in SKH-1 hairless mice. Topical application of NO-sulindac reduced tumor incidence, number (p<0.05) and volume (p<0.005) as compared to UVB (alone)-irradiated vehicle-treated mice. An increase in TUNEL-positive cells in skin lesions was accompanied by the enhanced Bax:Bcl-2 ratio. The expression of pro-apoptotic Bax was increased whereas anti-apoptotic Bcl-2 reduced. However, proliferation was identified as the major target of NO-sulindac in this study. A reduced expression of PCNA and cyclin D1 associated with the dampening of cell cycle progression was observed. The mechanism of this inhibition was related to the reduction in UVB-induced Notch signaling pathway. UVB-induced inflammatory responses were diminished by NO-sulindac as observed by a remarkable reduction in the levels of phosphorylated MAP Kinases Erk1/2, p38 and JNK1/2. In this regard, NO-sulindac also inhibited NFκB by enhancing IκBα as evidenced by the reduced expression of iNOS and COX-2, the direct NFκB transcription target proteins. NO-sulindac significantly diminished the progression of benign lesions to invasive carcinomas by suppressing the tumor aggressiveness and retarding epithelial-mesenchymal transition. A marked decrease in the expression of mesenchymal markers such as Fibronectin, N-cadherin, SNAI, Slug and Twist and an increase in epithelial cell polarity marker E-cadherin were noted in NO-sulindac-treated tumors. Our data suggest that NO-sulindac is a potent inhibitor of UVB-induced skin carcinogenesis and acts by targeting proliferation-regulatory pathways.


Assuntos
Antineoplásicos/uso terapêutico , Óxido Nítrico/metabolismo , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/prevenção & controle , Sulindaco/uso terapêutico , Raios Ultravioleta/efeitos adversos , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Pelados , Sulindaco/farmacologia
12.
Toxicol Appl Pharmacol ; 266(2): 233-44, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23147569

RESUMO

Histone deacetylase (HDAC) inhibitors are potent anticancer agents and show efficacy against various human neoplasms. Vorinostat is a potent HDAC inhibitor and has shown potential to inhibit growth of human xenograft tumors. However, its effect on the growth of skin neoplasm remains undefined. In this study, we show that vorinostat (2 µM) reduced expression of HDAC1, 2, 3, and 7 in epidermoid carcinoma A431 cells. Consistently, it increased acetylation of histone H3 and p53. Vorinostat (100mg/kg body weight, IP) treatment reduced human xenograft tumor growth in highly immunosuppressed nu/nu mice. Histologically, the vorinostat-treated tumor showed features of well-differentiation with large necrotic areas. Based on proliferating cell nuclear antigen (PCNA) staining and expression of cyclins D1, D2, E, and A, vorinostat seems to impair proliferation by down-regulating the expression of these proteins. However, it also induced apoptosis. The mechanism by which vorinostat blocks proliferation and makes tumor cells prone to apoptosis, involved inhibition of mTOR signaling which was accompanied by reduction in cell survival AKT and extracellular-signal regulated kinase (ERK) signaling pathways. Our data provide a novel mechanism-based therapeutic intervention for cutaneous squamous cell carcinoma (SCC). Vorinostat may be utilized to cure skin neoplasms in organ transplant recipient (OTR). These patients have high morbidity and surgical removal of these lesions which frequently develop in these patients, is difficult.


Assuntos
Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Neoplasias Cutâneas/tratamento farmacológico , Animais , Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas/patologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ciclinas/genética , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Feminino , Inibidores de Histona Desacetilases/administração & dosagem , Histona Desacetilases/genética , Humanos , Ácidos Hidroxâmicos/administração & dosagem , Hospedeiro Imunocomprometido , Camundongos , Camundongos Nus , Antígeno Nuclear de Célula em Proliferação/análise , Transdução de Sinais/efeitos dos fármacos , Neoplasias Cutâneas/patologia , Serina-Treonina Quinases TOR/metabolismo , Vorinostat , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Mol Cell Biochem ; 372(1-2): 249-56, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23001870

RESUMO

The nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ) plays a central role in regulating metabolism, including interaction with the estrogen receptor-α (ERα). Significantly, PPARγ activity can be modulated by small molecules to control cancer both in vitro and in vivo (Yin et al., Cancer Res 69:687-694, 2009). Here, we evaluated the effects of the PPARγ agonist GW7845 and the PPARγ antagonist GW9662 on DMBA-induced mammary alveolar lesions (MAL) in a mouse mammary organ culture. The results were as follows: (a) the incidence of MAL development was significantly inhibited by GW 7845 and GW 9662; (b) GW9662 but not GW7845, in the presence of estradiol, induced ER and PR expression in mammary glands and functional ERα in MAL; (c) while GW9662 inhibited expression of adipsin and ap2, GW 7845 enhanced expression of these PPARγ-response genes; and (d) Tamoxifen caused significant inhibition of GW9662 treated MAL, suggesting that GW9662 sensitizes MAL to antiestrogen treatment, presumably through rendering functional ERα and induction of PR. The induction of ERα by GW9662, including newer analogs, may permit use of anti-ER strategies to inhibit breast cancer in ER- patients.


Assuntos
Anilidas/farmacologia , Anticarcinógenos/farmacologia , Receptor alfa de Estrogênio/metabolismo , Glândulas Mamárias Animais/metabolismo , PPAR gama/antagonistas & inibidores , 9,10-Dimetil-1,2-benzantraceno , Animais , Sinergismo Farmacológico , Estradiol/fisiologia , Receptor alfa de Estrogênio/genética , Feminino , Glândulas Mamárias Animais/efeitos dos fármacos , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Oxazóis/farmacologia , PPAR gama/agonistas , PPAR gama/metabolismo , Lesões Pré-Cancerosas/induzido quimicamente , Lesões Pré-Cancerosas/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Tamoxifeno/farmacologia , Técnicas de Cultura de Tecidos , Ativação Transcricional/efeitos dos fármacos , Tirosina/análogos & derivados , Tirosina/farmacologia
14.
Exp Cell Res ; 317(10): 1359-67, 2011 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-21406194

RESUMO

Nitric oxide-donating aspirin (NO-ASA) is a promising agent for cancer prevention. Although studied extensively, its molecular targets and mechanism of action are still unclear. S-nitrosylation of signaling proteins is emerging as an important regulatory mechanism by NO. Here, we examined whether S-nitrosylation of the NF-κB, p53, and Wnt signaling proteins by NO-ASA might explain, in part, its mechanism of action in colon cancer. NO-ASA releases significant amounts of NO detected intracellularly in HCT116 and HT-29 colon cells. Using a modified biotin switch assay we demonstrated that NO-ASA S-nitrosylates the signaling proteins p53, ß-catenin, and NF-κB, in colon cancer cells in a time- and concentration-dependent manner. NO-ASA suppresses NF-κB binding to its cognate DNA oligonucleotide, which occurs without changes in the nuclear levels of the NF-κB subunits p65 and p50 and is reversed by dithiothreitol that reduces -S-NO to -SH. In addition to S-nitrosylation, we documented both in vitro and in vivo widespread nitration of tyrosine residues of cellular proteins in response to NO-ASA. Our results suggest that the increased intracellular NO levels following treatment with NO-ASA modulate cell signaling by chemically modifying key protein members of signaling cascades. We speculate that S-nitrosylation and tyrosine nitration are responsible, at least in part, for the inhibitory growth effect of NO-ASA on cancer cell growth and that this may represent a general mechanism of action of NO-releasing agents.


Assuntos
Aspirina/análogos & derivados , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , NF-kappa B/metabolismo , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico/metabolismo , Animais , Aspirina/farmacologia , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Técnicas Imunoenzimáticas , Masculino , Camundongos , Camundongos Nus , Tirosina/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , beta Catenina/metabolismo
15.
Mutat Res ; 746(1): 78-88, 2012 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-22498038

RESUMO

The genotoxic activities of three cancer chemopreventive drug candidates, CP-31398 (a cell permeable styrylquinazoline p53 modulator), SHetA2 (a flexible heteroarotinoid), and phospho-ibuprofen (PI, a derivative of ibuprofen) were tested. None of the compounds were mutagenic in the Salmonella/Escherichia coli/microsome plate incorporation test. CP-31398 and SHetA2 did not induce chromosomal aberrations (CA) in Chinese hamster ovary (CHO) cells, either in the presence or absence of rat hepatic S9 (S9). PI induced CA in CHO cells, but only in the presence of S9. PI, its parent compound ibuprofen, and its moiety diethoxyphosphoryloxybutyl alcohol (DEPBA) were tested for CA and micronuclei (MN) in CHO cells in the presence of S9. PI induced CA as well as MN, both kinetochore-positive (Kin+) and -negative (Kin-), in the presence of S9 at ≤100µg/ml. Ibuprofen was negative for CA, positive for MN with Kin+ at 250µg/ml, and positive for MN with Kin- at 125 and 250µg/ml. DEPBA induced neither CA nor MN at ≤5000µg/ml. The induction of chromosomal damage in PI-treated CHO cells in the presence of S9 may be due to its metabolites. None of the compounds were genotoxic, in the presence or absence of S9, in the GADD45α-GFP Human GreenScreen assay and none induced MN in mouse bone marrow erythrocytes.


Assuntos
Anticarcinógenos/toxicidade , Cromanos/toxicidade , Dano ao DNA/efeitos dos fármacos , Ibuprofeno/análogos & derivados , Mutagênicos/toxicidade , Organofosfatos/toxicidade , Pirimidinas/toxicidade , Tionas/toxicidade , Animais , Células CHO , Aberrações Cromossômicas , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Feminino , Humanos , Ibuprofeno/toxicidade , Camundongos , Testes para Micronúcleos , Testes de Mutagenicidade/métodos , Ratos , Ratos Endogâmicos F344 , Salmonella typhimurium/genética
16.
Gastroenterology ; 139(4): 1320-32, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20600034

RESUMO

BACKGROUND & AIMS: Nonsteroidal anti-inflammatory drugs (NSAIDs) are effective cancer chemopreventive agents. However, chronic administration of NSAIDs is associated with significant side effects, mainly of the gastrointestinal tract. Given these limitations, we synthesized phospho-sulindac (P-S; OXT-328), a novel sulindac derivative. METHODS: Here, we evaluated the safety and efficacy of P-S in preclinical models, including its mechanism of action with human colon cancer cell (HCCC) lines and animal tumor models. RESULTS: (1) Compared with sulindac, P-S is much more potent in inhibiting the growth of cultured HCCCs and more efficacious in preventing the growth of HT-29 xenografts in nude mice. P-S also prevents the growth of intestinal tumors in Apc/Min mice. (2) In combination with difluoromethylornithine (DFMO), P-S reduced tumor multiplicity in Apc/Min mice by 90%. (3) P-S is much safer than sulindac as evidenced by its in vitro toxicologic evaluation and animal toxicity studies. Mechanistically, P-S increases the intracellular levels of reactive oxygen and nitrogen species, which are key early mediators of its chemopreventive effect. Moreover, P-S induces spermidine/spermine N(1)-acetyltransferase enzymatic activity, and together with DFMO it reduces polyamine levels in vitro and in vivo. CONCLUSIONS: P-S displays considerable safety and efficacy, two pharmacologic properties that are essential for a potential cancer chemopreventive agent, and thus merits further evaluation.


Assuntos
Antineoplásicos/farmacologia , Neoplasias do Colo/prevenção & controle , Compostos Organofosforados/farmacologia , Sulindaco/análogos & derivados , Acetiltransferases/biossíntese , Animais , Poliaminas Biogênicas/análise , Linhagem Celular Tumoral , Dinoprostona/biossíntese , Eflornitina/farmacologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Sulindaco/farmacologia
17.
Biochem Biophys Res Commun ; 408(3): 363-8, 2011 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-21333626

RESUMO

Cyclosporine A (CsA) is an immunosuppressive drug commonly used for maintaining chronic immune suppression in organ transplant recipients. It is known that patients receiving CsA manifest increased growth of aggressive non-melanoma skin cancers. However, the underlying mechanism by which CsA augments tumor growth is not fully understood. Here, we show that CsA augments the growth of A431 epidermoid carcinoma xenograft tumors by activating tumor growth factor ß-activated kinase1 (TAK1). The activation of TAK1 by CsA occurs at multiple levels by kinases ZMP, AMPK and IRAK. TAK1 forms heterodimeric complexes with TAK binding protein 1 and 2 (TAB1/TAB2) which in term activate nuclear factor κB (NFκB) and p38 MAP kinase. Transcriptional activation of NFκB is evidenced by IKKß-mediated phosphorylation-dependent degradation of IκB and consequent nuclear translocation of p65. This also leads to enhancement in the expression of its transcriptional target genes cyclin D1, Bcl2 and COX-2. Similarly, activation of p38 leads to enhanced inflammation-related signaling shown by increased phosphorylation of MAPKAPK2 and which in turn phosphorylates its substrate HSP27. Activation of both NFκB and p38 MAP kinase provide mitogenic stimuli to augment the growth of SCCs.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Carcinógenos/farmacologia , Carcinoma de Células Escamosas/enzimologia , Ciclosporina/farmacologia , Imunossupressores/farmacologia , MAP Quinase Quinase Quinases/biossíntese , Animais , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Feminino , Humanos , Camundongos , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto
18.
J Pharmacol Exp Ther ; 337(3): 876-86, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21422165

RESUMO

We have developed a novel chemical modification of conventional nonsteroidal anti-inflammatory drugs to reduce their toxicity and enhance their efficacy. Phospho-ibuprofen [(PI) 2-(4-isobutyl-phenyl)-propionic acid-4-(diethoxy-phosphoryloxy)-butyl ester (MDC-917)], a novel derivative of ibuprofen, strongly inhibited the growth of human colon cancer cells in vitro and SW480 human colon cancer xenografts in nude mice. PI was metabolized minimally by cultured cells, but extensively by liver microsomes and mice, undergoing regioselective oxidation to produce 1-OH-PI and carboxyl-PI, which can be hydrolyzed to 1-OH-ibuprofen and carboxyl-ibuprofen, respectively. PI also can be hydrolyzed to release ibuprofen, which can generate 2-OH-ibuprofen, carboxyl-ibuprofen, and ibuprofen glucuronide. After a single oral administration (400 mg/kg) of PI, ibuprofen and ibuprofen glucuronide are the main plasma metabolites of PI; they have, respectively, C(max) of 530 and 215 µM, T(max) of 1 and 2 h, elimination t(1/2) of 7.7 and 5.3 h, and area under the concentration-time curve (0-24 h) of 1816 and 832 µM × h. Intact PI was detected in several tissues but not in plasma; at a higher PI dose (1200 mg/kg), PI plasma levels were 12.4 µM. PI generated the same metabolites in mouse plasma as conventional ibuprofen, but with much lower levels, perhaps accounting for the enhanced safety of PI. The antitumor effect of PI was significantly associated with plasma ibuprofen levels (p = 0.016) but not with xenograft ibuprofen levels (p = 0.08), suggesting a complex anticancer effect. These results provide a pharmacological basis to explain, at least in part, the anticancer efficacy and safety of this promising compound and indicate that PI merits further evaluation as an anticancer agent.


Assuntos
Antineoplásicos/farmacologia , Neoplasias do Colo/tratamento farmacológico , Ibuprofeno/análogos & derivados , Organofosfatos/farmacologia , Animais , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Neoplasias do Colo/metabolismo , Esterases/metabolismo , Feminino , Células HT29 , Humanos , Ibuprofeno/química , Ibuprofeno/metabolismo , Ibuprofeno/farmacocinética , Ibuprofeno/farmacologia , Camundongos , Camundongos Nus , Microssomos Hepáticos/metabolismo , Organofosfatos/química , Organofosfatos/metabolismo , Organofosfatos/farmacocinética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Mol Carcinog ; 50(7): 516-27, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21308804

RESUMO

Organ transplant recipients (OTRs) develop multiple aggressive and metastatic non-melanoma skin cancers (NMSCs). Yet, the underlying mechanism remains elusive. Employing a variety of immune-compromised murine models, immunoblotting, immunohistochemical and immunofluorescence techniques, we show that human squamous xenograft tumors in nude mice grow faster and become significantly larger in size following treatment with the immunosuppressive drug, cyclosporine A (CsA). Re-injected tumor cells isolated from CsA-treated xenografts continued to form larger tumors in nude mice than those from vehicle-controls and retained the CsA-signatures of calcineurin signaling inhibition. Similar results were obtained when these tumors were grown in SCID-beige mice or in immuno-competent mice inoculated with syngeinic tumor cells. Consistently, tumors in the CsA group manifested enhanced cellular proliferation and decreased apoptosis. Tumors in CsA-treated animals also showed an augmented epithelial-mesenchymal transition (EMT) characterized by an increased expression of fibronectin, α-SMA, vimentin, N-cadherin, MMP-9/-2, snail and twist with a concomitant decrease in E-cadherin. CsA-treated xenograft tumors manifested increased TGFß1 expression and TGFß-dependent signaling characterized by increased nuclear p-Smad 2/3. Our data demonstrate that CsA alters the phenotype of skin SCCs to an invasive and aggressive tumor-type by enhancing expression of proteins regulating EMT acting through the TGFß1 signaling pathway providing at least one unique mechanism by which multiple aggressive and metastatic NMSCs develop in OTRs.


Assuntos
Carcinoma de Células Escamosas/patologia , Ciclosporina/farmacologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transdução de Sinais , Neoplasias Cutâneas/patologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Carcinoma de Células Escamosas/metabolismo , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Imunofluorescência , Humanos , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Nus , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Cutâneas/metabolismo , Transplante Heterólogo
20.
Arch Biochem Biophys ; 508(2): 159-63, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21232524

RESUMO

Nonmelanoma skin cancer (NMSC) is the most common human cancer, with an incidence of more than 1.2 million per year in the USA. The risk for the development of NMSCs increases by approximately 10-250 fold in chronically immune suppressed organ transplant recipients (OTRs). Solar UVB is the most common etiologic factor in the development of this neoplasm, both in immune competent and immune suppressed populations. This review provides a description of NMSC in OTRs. It also provides an account of the various immunologic and non-immune-dependent mechanisms involved in the pathogenesis and progression of NMSCs in OTRs. Finally, this review addresses possible strategies for the prevention of this cancer, particularly focusing on the aspects that may be incorporated to prevent negative effects of chemopreventive chemicals on graft survival.


Assuntos
Imunossupressores/efeitos adversos , Transplante de Órgãos/efeitos adversos , Neoplasias Cutâneas/patologia , Animais , Inibidores de Ciclo-Oxigenase/farmacologia , Humanos , Inibidores da Ornitina Descarboxilase , Retinoides/farmacologia , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA